Você está na página 1de 13

Received: 7 August 2022 Revised: 28 January 2023 Accepted: 3 February 2023

DOI: 10.1002/glia.24353

REVIEW ARTICLE

The role of glial cells in Zika virus-induced neurodegeneration

André Quincozes-Santos 1,2,3 | Larissa Daniele Bobermin 2 |


Naithan Ludian Fernandes Costa 2 | Natalie K. Thomaz 3 |
Rômulo Rodrigo de Souza Almeida 3 | Walter O. Beys-da-Silva 4 | Lucélia Santi 4 |
Rafael L. Rosa 4 | Daniela Capra 5 | Juliana M. Coelho-Aguiar 6 |
Marcos Fabio DosSantos 7 | Manoela Heringer 5 | Elizabeth O. Cirne-Lima 4 |
Jorge Almeida Guimarães 4 | Lavínia Schuler-Faccini 4 |
Carlos-Alberto Gonçalves 1,2,3 | Vivaldo Moura-Neto 5,6 | Diogo Onofre Souza 1,3
1
Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
2
 s-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
Programa de Po
3
 s-Graduação em Ciências Biolo
Programa de Po gicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre,
RS, Brazil
4
Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
5
Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
6
 rio de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
Laborato
7
 rio de Propriedades Mecânicas e Biologia Celular, Faculdade de Odontologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
Laborato

Correspondence
André Quincozes-Santos, Laborato rio de Abstract
Neurotoxicidade e Glioproteção (LABGLIO),
Zika virus (ZIKV) is a strongly neurotropic flavivirus whose infection has been associ-
Departamento de Bioquímica, Universidade
Federal do Rio Grande do Sul, Rua Ramiro ated with microcephaly in neonates. However, clinical and experimental evidence
Barcelos, 2600 – Anexo, Bairro Santa Cecília.
indicate that ZIKV also affects the adult nervous system. In this regard, in vitro and
90035–003 Porto Alegre, RS, Brazil.
Email: andrequincozes@ufrgs.br in vivo studies have shown the ability of ZIKV to infect glial cells. In the central ner-
vous system (CNS), glial cells are represented by astrocytes, microglia, and oligoden-
Funding information
Conselho Nacional de Desenvolvimento drocytes. In contrast, the peripheral nervous system (PNS) constitutes a highly
gico; Coordenação de
Científico e Tecnolo
heterogeneous group of cells (Schwann cells, satellite glial cells, and enteric glial cells)
Aperfeiçoamento Pessoal de Nível Superior
(CAPES); Fundação de Amparo à Pesquisa do spread through the body. These cells are critical in both physiological and pathological
Estado do Rio Grande do Sul (FAPERGS);
conditions; as such, ZIKV-induced glial dysfunctions can be associated with the devel-
Ministry of Science, Technology, Innovation
and Communications (MCTIC), Ministry of opment and progression of neurological complications, including those related to the
Education (MEC), Ministry of Health (MS),
adult and aging brain. This review will address the effects of ZIKV infection on CNS
Grant/Award Numbers: Edital MCTIC/
FNDCT-CNPq/MEC-CAPES/MS-Decit/No and PNS glial cells, focusing on cellular and molecular mechanisms, including changes
14/2016, project 440763/2016-9
in the inflammatory response, oxidative stress, mitochondrial dysfunction, Ca2+ and
glutamate homeostasis, neural metabolism, and neuron–glia communication. Of note,
preventive and therapeutic strategies that focus on glial cells may emerge to delay
and/or prevent the development of ZIKV-induced neurodegeneration and its
consequences.

André Quincozes-Santos and Larissa Daniele Bobermin contributed equally to this work.

Glia. 2023;71:1791–1803. wileyonlinelibrary.com/journal/glia © 2023 Wiley Periodicals LLC. 1791


10981136, 2023, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/glia.24353 by UNSA - Univ Nacional de Salta, Wiley Online Library on [06/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1792 QUINCOZES-SANTOS ET AL.

KEYWORDS
glial cells, inflammatory response, neurodegeneration, neuron–glia communication, Zika virus

1 | I N T RO DU CT I O N were documented in children, and the virus was identified as a human


teratogen and associated with clinical manifestations, including micro-
In 2016, the World Health Organization declared Zika virus (ZIKV) cephaly, which were collectively named congenital Zika syndrome
infection as a ‘Public Health Emergency of International Interest”, not (CZS) (Schuler-Faccini et al., 2022). According to a database of imaging
only because of the high risk of disease transmission, but also due to studies, cranial characteristics of children with microcephaly were
the neurological consequences that began to be observed on a large described, including a substantial increase in ventricle volumes, corti-
scale, in children and adults. ZIKV is an arbovirus that was first identi- cal and subcortical atrophy, cerebellar hypoplasia and abnormalities in
fied and isolated in 1947 in Rhesus monkeys from the Zika forest of the white matter, corpus callosum, basal ganglia, and trunk, as well as
Uganda. This virus belongs to the family Flaviviridae and is mainly calcifications in different brain regions (Schuler-Faccini et al., 2022;
transmitted by Aedes mosquitoes. ZIKV was endemic, with few cases Wu et al., 2018). Moreover, diagnosis of autism spectrum disorders
reported in humans in Africa and Asia until 2007, when its geographic (ASD) in childhood were recently correlated with ZIKV infection dur-
distribution spread, reaching the Yap Islands (in 2007), French Polyne- ing pregnancy (Santi et al., 2021; Vianna et al., 2018).
sia (in 2013), and the Americas (in 2015) (Musso et al., 2019). In addition, current clinical data show that ZIKV infection affects
ZIKV can infect several body sites, including the eyes, cardiac the adult brain, causing neurological complications, including Guillain-
tissue, the gastrointestinal (GI) system, reproductive tract, placenta, Barré, seizures, and other myelitis and encephalitis-like illnesses
and central nervous system (CNS) (Figure 1) (Gasco & Muñoz- (Carteaux et al., 2016; Ferrari-Marinho et al., 2022; Henriques-Souza
Fernández, 2020; Rossi et al., 2020; Ryan et al., 2021; Zafar et al., 2017). Neuroimaging findings indicate changes in the adult
et al., 2021). The most common symptoms of acute ZIKV infection human brain structure and functional organization, comprising both
include fever, headaches, myalgias, rash, and conjunctivitis. Less fre- motor-related and nonsomatomotor regions (Bido-Medina
quent symptoms have also been reported, such as diarrhea, edema, et al., 2018; Ferrari-Marinho et al., 2022). Furthermore, accumulating
and retro-orbital eye pain (Petersen et al., 2016; Rawal et al., 2016). experimental evidence has demonstrated the ability of ZIKV to infect
After the ZIKV outbreak in Brazil at 2015, neurological symptoms neural cells and replicate in adult brain tissue, being able to affect

F I G U R E 1 Schematic illustration of Zika virus (ZIKV) infection in body organs, pregnant women, and children. ZIKV can infect several cell
types, including eye, cardiac, gastrointestinal, reproductive tract, placenta, and central nervous system cells (a). In pregnant women, viral
transmission can occur through the placenta (b). Due to ZIKV neurotropism, infections can impair brain development in infants/children, even in
the absence of evident structural brain abnormalities and microcephaly (c).
10981136, 2023, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/glia.24353 by UNSA - Univ Nacional de Salta, Wiley Online Library on [06/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
QUINCOZES-SANTOS ET AL. 1793

synapses and induce cognitive deficits (Figueiredo et al., 2019). The et al., 2017; Liang et al., 2016). In vitro and in vivo experimental
areas of most significant infection were reported to be the regions of models indicate that pathological mechanisms of CZS, as well as the
cognitive processing and memory (frontal cortex and hippocampus) progression and clinical outcomes of ZIKV infection, are closely asso-
and motor area (striatum) (Figueiredo et al., 2019). ZIKV has also been ciated with neuron–glia communication (Beys-da-Silva et al., 2020;
detected in cerebrospinal fluid (CSF) and the brain tissue of children Bobermin et al., 2020; Greenhalgh et al., 2020; Ledur et al., 2020). In
with CZS, and in adult patients with neurological complications addition, glial cells have a wide range of adaptive functions that regu-
(Alves-Leon et al., 2019; Figueiredo et al., 2019; Mlakar et al., 2016; late tissue recovery after injury, since dysfunctions in these cells may
Schuler-Faccini et al., 2022), reinforcing its neurotropism. represent a risk factor for neurodegenerative and neuropsychiatric
It is important to note that although ZIKV infection can be tran- diseases, and consequently, for ZIKV-induced neurodegeneration
sient, it can induce significant changes in adult brain functionality with (Labzin et al., 2018; Li et al., 2021; Mehta et al., 2013).
potential impact on the pathogenesis of neurodegenerative diseases, Additionally, there is a close relationship between ZIKV and glial
which mostly involve glial cells. Thus, in this review, we will discuss cells whereby; (i) astrocytes present the AXL receptor, a candidate
the role of glial cells as central players in the neurodegenerative com- receptor in neural cells for promoting ZIKV infection (Chen
plications associated with ZIKV infection. et al., 2018; Meertens et al., 2017), and roles of astrocytes in produc-
tive infection and possibly as a ZIKV reservoir have been demon-
strated (Huang et al., 2018; Stefanik et al., 2018); (ii) oligodendrocytes
2 | G LI A L C EL L S : OR I G I N S A N D are the myelin-producing cells that insulate axons to allow saltatory
FUNCTIONS conduction; previous findings have reported their susceptibility to
ZIKV infection (Musso et al., 2019; Schultz, Cumberworth,
The term “glial cells”, or the concept of “neuroglia,” is based on the et al., 2021), and ZIKV-induced impairment in OPC differentiation and
collective function that these types of cells play in nervous system oligodendrocyte development (Li, Wang, et al., 2018); (iii) microglia
homeostasis, regardless of their distinct structure, specific functions, are the CNS resident immune cells, are proposed to be targets of
and embryonic origins (Verkhratsky et al., 2019). Glial cells were previ- ZIKV, and mediate neuroinflammation after ZIKV exposure (Meertens
ously mischaracterized as a mere sort of “glue” for adjoining neurons et al., 2017; Rombi et al., 2020); and (iv) Guillain-Barré syndrome,
without further, more complex functions (García-Marín et al., 2007; which is related to peripheral glial cells, has been described during
Jäkel & Dimou, 2017). From this concept arose the name glia (from ZIKV outbreaks (Musso et al., 2019; Rombi et al., 2020). Figure 2 sum-
the ancient Greek word “glía” and the German word “Nervenkitt”, marizes the main effects of ZIKV in glial cells, which are addressed in
both meaning “glia” in English). However, several functions have since the following sections.
been attributed to glial cells, including synapse formation, regulation
of synaptic transmission and plasticity, redox balance, ion and water
homeostasis, maintenance of the blood–brain barrier (BBB), control of 3.1 | Effects of ZIKV on glial metabolism, oxidative
the toxicity formed in the extracellular space, and the establishment stress, and glutamate homeostasis
of myelin sheets (Jäkel & Dimou, 2017).
In addition, glial cells not only act in healthy states, but are also Astrocytes contain numerous stellate processes, due to their specific
crucial in pathological conditions, a topic that has been extensively morphology and intimate contact with synapses, blood vessels, and
studied. In this regard, glial cells display immune and inflammatory other glial cells. ZIKV infection triggers morphological changes in
functions, which affect the progression and neurological conse- astrocytes (Huang et al., 2018; Stefanik et al., 2018). In addition,
quences of virus infections (Inglis et al., 2016; Li, Wang, et al., 2018; astrocytes support neurons through metabolic coupling between syn-
Meertens et al., 2017). Glial cells are found in the CNS, where they aptic activity and glucose utilization (neurometabolic coupling)
are represented by microglia, astrocytes, oligodendrocytes and oligo- (Bélanger et al., 2011), a classic example of neuron–glia interaction.
dendrocyte progenitor cells (OPCs), and in the peripheral nervous sys- The metabolic activity of neural cells can be reprogrammed in
tem (PNS), where they are composed of a highly heterogeneous response to redox and inflammatory challenges (Lynch, 2020); more-
group of cells spread through the body. Peripheral glial cells include over, increased glucose consumption may be needed for viral replica-
Schwann cells, satellite glial cells, and enteric glial cells, among others tion, leading to energy stress in brain cells. It has been hypothesized
(Mai & Paxinos, 2012). that ZIKV replication may increase glucose uptake through the
GLUT-1 glucose transporter, similar to Dengue virus (Fontaine
et al., 2015; Gilbert-Jaramillo et al., 2019). While increased glycolysis
3 | GLIAL CELLS AS TARGETS OF ZIKV produces ATP rapidly for viral replication, it also contributes to endo-
INFECTION plasmic reticulum stress, mitochondrial dysfunction, elevated reactive
oxygen species (ROS) production and dysregulated autophagy, which
ZIKV infection strongly affects the cytoarchitecture of the brain and lead to cell death (Gilbert-Jaramillo et al., 2019). In immune cells, gly-
specific neurochemical and molecular functions of the neural cells, colytic enzymes also trigger hypoxia inducible factor 1α (HIF1α), the
including glial fibrillary acidic protein (GFAP) positive cells (Devhare production of interleukin-1β (IL-1β) and inflammasome activation
10981136, 2023, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/glia.24353 by UNSA - Univ Nacional de Salta, Wiley Online Library on [06/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1794 QUINCOZES-SANTOS ET AL.

neurotransmitter in the mammalian CNS. However, an excessive con-


centration of glutamate in the synaptic cleft is implicated in the patho-
genesis of several brain disorders, a phenomenon termed
excitotoxicity (Allaman et al., 2011). Astrocytes are responsible for the
glutamate uptake and the regulation of extracellular levels of gluta-
mate. The impairment of glutamate transporters causes excitotoxicity,
which is potentiated by Ca2+ release from the mitochondria and
endoplasmic reticulum. In iPSC-derived astrocytes, ZIKV interfered
with Ca2+ uptake by mitochondria, increasing oxidative stress and cell
damage (Ledur et al., 2020). It is noteworthy that brain disorders share
common pathomechanisms (Lewerenz & Maher, 2015; Mehta
et al., 2013), therefore, changes in glial metabolism, redox and gluta-
mate homeostasis induced by ZIKV can impair the adult brain and
contribute to further neurological manifestations related to ZIKV
infection.

3.2 | Induction of glial cell-mediated


neuroinflammation by ZIKV

F I G U R E 2 Zika virus (ZIKV)-induced glial dysfunctions potentially


Astrocytes and microglia have a critical role in brain pathophysiology
associated with neurodegeneration. ZIKV infection can induce several
functional changes in glial cells, including inflammatory responses, and influence immunological processes and events caused by ZIKV
oxidative stress, and mitochondrial dysfunction, as well as alterations infection (Rombi et al., 2020). These glial cells synthesize and release
in Ca2+ and glutamate homeostasis, metabolism, and neuron–glia several cytokines and chemokines (Vainchtein & Molofsky, 2020),
communication. These changes may converge to cause where the inflammatory process constitutes a primary event connect-
neurodegeneration. Functional glial cells are represented by ramified
ing ZIKV infection and glial cells. ZIKV triggers innate antiviral immune
astrocyte (blue) and microglia (yellow), and oligodendrocyte in purple.
responses, such as activation of the NLR family pyrin domain contain-
Dysfunctional glial cells are represented by astrocyte (blue) and
microglia (yellow) with enlarged cell bodies, and oligodendrocytes in ing 3 (NLRP3) inflammasome and subsequent release of IL-1β in glial
gray. Functional (left) and damaged (right) neurons are represented in cells, inducing cell death (Tricarico et al., 2017). Moreover, ZIKV infec-
the center, and ZIKV is represented by yellow circles. tion increases the expression of Toll-like receptors (TLR2 and TLR3),
which in turn can mediate viral-induced inflammatory response and
replication in astrocytes (Bobermin et al., 2020; Ojha et al., 2019).
(Hughes & O'Neill, 2018; Palsson-McDermott et al., 2015), while inhi- Interestingly, prolonged infection of astrocytes may lead to the spread
bition of the glycolytic pathway can revert neuroinflammation of infection to other cell types, inducing inflammation and further
(Vizuete et al., 2022). damage, even in uninfected cells, due to astrocyte loss (Retallack
It is also important to emphasize that an experimental in vitro et al., 2016). In contrast, astrocytes are suggested to have a role in
model of ZIKV infection was found to present significant mitochon- limiting the spread of ZIKV, since infection of primary human astro-
drial failure, oxidative stress, and DNA damage in human iPSC-derived cytes induced the production of type I interferon, preventing the
astrocytes (Ledur et al., 2020). In the glial cell line, U87-MG, ZIKV virus-induced killing of the cells (Lindqvist et al., 2016). In addition,
infection induced an increase in the production of ROS, lipid peroxida- astrocytes form functional barriers such as the BBB, which restrict the
tion, and protein carbonylation, in addition to decreased activities of entry of inflammatory mediators into the CNS (Greenhalgh
the catalase (CAT) and the superoxide dismutase (SOD) enzymes, in et al., 2020). However, this function can be compromised by ZIKV
association with a decrease in nuclear factor erythroid 2-related factor infection since its pathophysiological and immunological features indi-
2 (Nrf2) activation (Almeida et al., 2020). Furthermore, oxidative cate that the cytokine storm may affect BBB permeability
stress, associated with antioxidant depletion, was found in the brain (Vainchtein & Molofsky, 2020). In line with this, the CSF contains a
of ZIKV-infected mice (Almeida et al., 2020). Thus, astrocytes are criti- variety of immune cells that are related to adaptive immunity, and
cal for controlling redox homeostasis, and changes in this function can ZIKV has also been detected in the CSF (França et al., 2016). Thus,
impair the adult brain, contributing to further neurological manifesta- astrocytes can contribute to propagating the development and pro-
tions related to ZIKV infection (Jiwaji & Hardingham, 2022; Ledur gression of ZIKV infection in the CNS, causing the injury of neural
et al., 2020; Schreiner et al., 2015). cells through direct infection-induced and/or indirect immune-
Dysfunctions in mitochondrial activity may lead to incorrect func- mediated mechanisms (Rombi et al., 2020).
tioning of specific astrocyte proteins, such as glutamate transporters Although microglial cells can adopt the inflammatory phenotype
(Allaman et al., 2011). Glutamate is the major excitatory to exert neuroprotective functions, the persistent activation of
10981136, 2023, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/glia.24353 by UNSA - Univ Nacional de Salta, Wiley Online Library on [06/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
QUINCOZES-SANTOS ET AL. 1795

microglia can be associated with chronic neuroinflammation, a fact Peripheral infection of adult knockout mice for the type-I inter-
related to neurodegenerative and neuropsychiatric disorders. In this feron receptor gene (A129 mice), which have an inefficient interferon
context, neuroinflammation is a mutual feature of congenital micro- response, are associated with high levels of viral RNA in all analyzed
cephaly in newborns and neurological complications in childhood and regions of the brain (Figueiredo et al., 2019). Upregulation of tumor
adults (Maucourant et al., 2019). Furthermore, ZIKV infection of necrosis factor α (TNF-α) and complement system proteins C1q and C3
microglial cells can inhibit cell proliferation and neuronal differentia- can impact neuron–glia communication, and consequently, the onset of
tion of neural precursor cells (Lum et al., 2017; Wang et al., 2018), as neurodegeneration (Figueiredo et al., 2019). In hippocampal slices from
well as microgliosis in the brain of adult mice, particularly in the hippo- adult rats, acute exposure to ZIKV caused significant cellular alterations
campus, suggesting a role for these cells in the control of ZIKV replica- regarding redox homeostasis, inflammatory processes, neurotrophic
tion and/or its elimination in the brain (Enlow et al., 2021; Figueiredo functions, glial commitment, and molecular signaling pathways, which
et al., 2019). Consistent with this hypothesis, experimental models of potentially affect important aspects of neuron–glia communication.
flavivirus infection, including ZIKV, can cause brain lesions in the hip- Moreover, ZIKV-induced neuroinflammation can involve Nrf2 and
pocampus of mice, where infiltrated macrophages and resident micro- nuclear factor kappa B (NFκB) pathways (Bobermin et al., 2020). Nrf2 is
glia are significantly increased. Moreover, T cell-derived interferon-γ a transcription factor involved in the adaptive responses to oxidative
(IFN-γ) signaling is required for microglial activation and seems to be stress and inflammation through their target genes. As such, there is an
involved in ZIKV post-infection cognitive sequelae by eliminating pre- interplay between Nrf2 and NFκB signaling, which may be critical
and post-synaptic terminals (Garber et al., 2019). Flaviviruses can also mechanistic partners in the altered neuron–glial communication
promote type 1 macrophage polarization in the brain, which is associ- observed after ZIKV exposure (Bobermin et al., 2020). Accordingly,
ated with encephalitic severity (Jhan et al., 2021). Other experimental NFκB can control energy homeostasis and metabolic adaptation by reg-
data from the infected brain of mice indicate that ZIKV with high ulating mitochondrial respiration, and reprogramming cellular bioener-
pathogenicity may induce sustained activation of the immune system, getic pathways toward glucose metabolism, which requires an
including macrophage activation, leading to nerve tissue damage orchestrated regulation between neurons and glial cells in the CNS
(Shang et al., 2022). ZIKV may also evade host innate immune (Mauro et al., 2011). In addition, NFκB triggers an inflammatory
response in macrophages and glial cells, which could facilitate viral response by increasing pro-inflammatory cytokines that may induce
replication and dissemination in these cells (Gim et al., 2019). There- persistent neuroinflammation (Lima et al., 2019). As such, it is vital to
fore, in cooperation with resident microglia, macrophage activation consider the link between metabolic and inflammatory signatures and
may facilitate CNS inflammation and neurotoxicity during ZIKV their consequences on neuron–glia coupling (Lynch, 2020). NFκB can
infection. also regulate glutamate transporter activity (Ghosh et al., 2011), which
ZIKV can also directly affect oligodendroglia, infecting glial pro- might link mitochondrial-mediated ROS overproduction, inflammatory
genitors during development, and disturbing oligodendrocyte func- response, and excitotoxicity. These events are the basis of a number of
tionality, accompanied by immune response and microglial activation neurological and functional disabilities, including Alzheimer's disease
(Li, Wang, et al., 2018; Schultz, Barrie, et al., 2021; Zhang et al., 2017). (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and
In mature myelinated CNS cultures, ZIKV infection damages myelin- multiple sclerosis, among others (Bhat et al., 2015; Labzin et al., 2018;
ated fibers (Cumberworth et al., 2017; Schultz, Barrie, et al., 2021). Of Verma et al., 2022), which may be associated with different clinical out-
note, oligodendrocyte cell death might lead to secondary autoimmune comes after a patient's recovery from acute ZIKV infection.
demyelination (Molina-Gonzalez et al., 2022), and these observations Astrocytes synthesize and release a wide range of signaling mole-
are critical for understanding the effects of ZIKV in the mature CNS. cules, including gliotransmitters, and can therefore affect other glial
cells and neurons through the gliotransmission process, which is
closely associated with the “gliocrine system” (Vardjan et al., 2019).
3.3 | ZIKV affects neuron–glia communication As such, the release of gliosignaling molecules (Vardjan et al., 2019)
can occur through vesicular exocytosis or by carrier-mediated trans-
ZIKV can affect numerous signaling pathways that play critical roles in port or channel flux through the plasma membrane, which seems to
the crosstalk between metabolism, inflammatory response, and oxida- be activated mainly under pathological conditions (Gundersen
tive systems, affecting neuron–glia communication. In astrocytes, et al., 2015; Vardjan & Zorec, 2015). Homeostatic imbalance and
ZIKV dysregulates gene expression and pathways associated with astroglial vesicular displacement contribute to the pathophysiology of
essential functions/processes, including metabolism and lipid biosyn- neurodegenerative diseases, including AD, Huntington's disease, and
thesis, glial differentiation, neurogenesis, and brain development, neuroinfections, such as ZIKV (Zorec et al., 2018).
along with pathways such as phosphoinositide-3-kinase (PI3K)/Akt Finally, the glial reactivity/glial activation observed in experimental
(Schouest et al., 2021; Shereen et al., 2021). These data agree with a models and studies with post-mortem brains (Bobermin et al., 2020;
previous study from our group that showed that ZIKV potentially Enlow et al., 2021; Figueiredo et al., 2019; Ledur et al., 2020) may eluci-
induces reprogramming of the cellular metabolic machinery date the effects of ZIKV infection on neuronal loss and neurological def-
and inhibits autophagy, neurogenesis, and immune response icit, emphasizing the role of neuron–glia communication in explaining
(Beys-da-Silva et al., 2019). the pathogenesis of CZS and other neurological diseases associated with
10981136, 2023, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/glia.24353 by UNSA - Univ Nacional de Salta, Wiley Online Library on [06/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1796 QUINCOZES-SANTOS ET AL.

ZIKV. Table 1 displays important publications investigating the effects (NC). ZIKV can productively infect human NC cells and peripheral
of ZIKV on CNS cells, particularly glial cells, in this review. neurons derived from human pluripotent stem cells (hPSCs), inducing
increased cell death and transcriptional dysregulation. The presence
of ZIKV in Schwann cells differentiated from hPSCs has also been
3.4 | The role of glial cells in ZIKV observed (Oh et al., 2017). Schwann cells are among the most impor-
infection-induced alterations in neurosensory tant peripheral glial cells. They form the myelin sheet, which sur-
pathways, peripheral and enteric nervous system rounds the axons of the peripheral nerves in a spiral manner
(Verkhratsky et al., 2019). ZIKV infection and replication has been
Previous studies conducted in mice demonstrated that ZIKV infection demonstrated in human Schwann cells, which may lead to myelin dis-
targets peripheral neural phenotypes derived from the neural crest ruption and peripheral neuropathies, such as Guillain-Barré syndrome

TABLE 1 Effects of Zika virus on glial cells

Experimental models Main findings Reference


In vitro
CNS and PNS myelinating cultures ZIKV infection is enhanced in the absence of the type I interferon; CNS axons Cumberworth
and myelinating oligodendrocytes are especially vulnerable, compared to PNS et al., 2017
cells
hPSC-derived astrocytes, U87 cells AXL blocking reduced ZIKV infection; azithromycin reduced viral proliferation Retallack et al., 2016
and organotypic culture and cytopathic effects
Human astrocytes AXL promotes ZIKV infection by antagonizing type I IFN signaling Chen et al., 2018
Human astrocytes AXL is important to ZIKV infection; ZIKV activated autophagy and inflammatory Ojha et al., 2019
responses by TLR pathway
Human astrocytes ZIKV infection through AXL modulates the innate response Meertens et al., 2017
Human astrocytes ZIKV specifically infects GFAP and S100B-positive astrocytes Huang et al., 2018
Human astrocytes Expression of cytokines/chemokines Stefanik et al., 2018
Human microglia Inflammatory response Lum et al., 2017
iPSC-derived astrocytes Glial reactivity, ROS imbalance, mitochondrial defects and DNA breakage Ledur et al., 2020
Mice astrocytes Astrocytes mediate a local IFN response within the CNS during flavivirus Lindqvist et al., 2016
infection.
Mouse astrocytes and human Dysregulation in the expression of pathways associated with cell death, Shereen et al., 2021
astroglioma metabolism, transcription, DNA replication and repair, cell cycle, and viral
responses
Murine embryonic spinal cord Signs of myelin damage, alongside axonal injury and diminished axonal density; Schultz, Barrie,
cell-derived and CNS myelinating upregulation of factors that have been linked to reduced myelination and et al., 2021
cultures demyelination
Rat hippocampal slices Inflammatory response, redox imbalance, modulation of neurotrophic factor Bobermin et al., 2020
release and changes in the expression of neuron-glial signaling pathways
Rhesus macaque astrocytes Induction of pathways associated with cell death, viral replication, metabolic and Schouest et al., 2021
lipid biosynthesis
In vivo
C57BL/6 mice Oxidative stress and decrease in antioxidant enzyme activities in vitro and in vivo Almeida et al., 2020
C57BL/6 J, Ifngr1 / , Ifngr f/fl and Neuroinflammation, neuronal loss in the hippocampus and microglial activation Garber et al., 2019
CX3CR1-CreERT mice
C57BL/6 mice, Swiss mice, Brain inflammation with microglial activation, phagocytosis of hippocampal Figueiredo et al., 2019
Il1r / mice and adult human synapses and memory impairment in mice; ZIKV replicates in adult human
cortical slices brain tissue
Ifnar1 knockout Apoptosis in early myelinating oligodendrocytes in white matter Schultz, Cumberworth,
et al., 2021
Mouse Neuronal cell death, abolishment of oligodendrocyte development, and immune Zhang et al., 2017
response

Note: In vitro and in vivo experimental models focusing on the effects of Zika virus on neuron-glial cellular and molecular mechanisms associated with
neurodegenerative diseases.
Abbreviations: BBB, blood–brain barrier; CNS, central nervous system; GFAP, glial fibrillary acidic protein; IFN, interferon; PNS, peripheral nervous system;
ROS, reactive oxygen species; TLR, toll-like receptors; ZIKV, Zika virus.
10981136, 2023, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/glia.24353 by UNSA - Univ Nacional de Salta, Wiley Online Library on [06/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
QUINCOZES-SANTOS ET AL. 1797

(Dhiman et al., 2019; Volpi et al., 2018). Schwann cells also escheat (Coulter & Eid, 2012; Labzin et al., 2018; Mattson &
the central processes of the spiral (cochlear) ganglion axons, a group Arumugam, 2018; Quincozes-Santos et al., 2021; Rudy et al., 2015).
of neuron cells located within the cochlea's modiolus (Carricondo & Therefore, it can be speculated that glial cells participate in the patho-
mez, 2019).
Romero-Go mechanisms associated with neurological complications related to
ZIKV can also change the auditory pathway (de Barbosa ZIKV infection in the adult and aging brain, since ZIKV can signifi-
et al., 2019; Inglis et al., 2016) and induce sensorineural hearing loss cantly change this triad.
(Leal et al., 2016), possibly due to a direct action in the cochlea, as also Although glutamate plays a vital role in learning and memory,
observed for other congenital viral infections. Therefore, it may be alterations in glutamatergic signaling can lead to excitotoxicity (Rudy
hypothesized that ZIKV may efficiently replicate along the glial cells et al., 2015). ZIKV may interfere with glutamate synapse mechanisms,
present in the auditory system, leading to hearing loss and CNS since it replicates in adult mice brain tissue and affects synapses and
effects/dysfunctions. In addition, ZIKV can replicate in the olfactory memory; therefore, cognitive deficits could be considered as a poten-
ensheathing cells, the glial cells associated with the olfactory system tial comorbidity in ZIKV-infected adults (Figueiredo et al., 2019). In
(Mutso et al., 2020). These cells also surround and support the axons addition, ZIKV infection early in life, in mice, has been associated with
of olfactory neurons throughout their trajectory from the olfactory long-term neuropathological and behavioral consequences, which is
mucosa to the olfactory bulb (Ekberg & St john, 2014). In this scenario, accompanied by a persistent ZIKV replication (de Nem et al., 2018;
the olfactory pathway represents a route that can be used by different Ireland et al., 2020). One of the possible factors associated with neu-
types of bacteria and viruses to reach the brain (St. John et al., 2016). ropathy is the levels of glutamate in astrocytes, which may result in
Remarkably, the findings that both olfactory ensheathing glial cells glutamatergic excitotoxicity through decreased glutamate transporter
and brain microvascular cells support ZIKV replication may relate to activity via ROS and inflammatory cytokines (Sirohi & Kuhn, 2017).
the ability of the virus to enter the CNS via the olfactory nerve or Downregulation of excitatory amino acid transporter 2 (EAAT2) and
through the BBB (Mutso et al., 2020). Moreover, it should be noted glutamine synthetase in astrocytes has been observed in a neuroinva-
that, in adults, sensory changes in auditory and olfactory pathways sive respiratory virus model (Brison et al., 2014); however, at least
may also precede symptoms of neurodegenerative diseases such as acutely, ZIKV exposure did not change the expression of astroglial
AD and PD (Shad et al., 2022; Ubeda-Bañon et al., 2020). glutamate transporters in hippocampal slices (Bobermin et al., 2020).
Although considered unusual and rarely mentioned in the litera- Further experimental investigations of how ZIKV can imbalance gluta-
ture, GI symptoms are among the most related to ZIKV infection mate levels are needed, although a direct excitotoxic mechanism of
(Bôtto-Menezes et al., 2019; Li, Deng, et al., 2018; Rawal et al., 2016). ZIKV infection, mediated by the N-methyl-D-aspartate (NMDA)
The injection of ZIKV into A129 mice was found to infect the myen- receptor, has been demonstrated in neuronal cultures (Costa
teric plexus of the small intestine (more specifically in the jejunum) et al., 2017; Gaburro et al., 2018). Moreover, ZIKV-infected neurons
(Oh et al., 2017), but little is known regarding the effects of ZIKV release increased levels of glutamate (Olmo et al., 2017). Interestingly,
infection on the GI tract and the enteric nervous system. The enteric NMDA blockers inhibited ZIKV replication and prevented neuronal
glia show great plasticity, with a high proliferation capacity and are death in in vitro and in vivo experimental models (Costa et al., 2017).
able to act as progenitor cells and differentiate into neurons in specific These findings suggest an important pathway underlying ZIKV
situations (Gershon, 2011; Veríssimo et al., 2019). In contrast to previ- induced neurological effects, since activation of NMDA receptors is a
ous evidence that did not identify enteric glia ZIKV infection determinant event for the excitotoxic process in neurodegenerative
(Martínez et al., 2020), our preliminary findings obtained in a cell line diseases.
of rat enteric glial cells show that the enteric glia may be persistently Astrocytes can also regulate extracellular glutamate levels
and productively infected with ZIKV (data not shown). However, the through hemichannels, volume-regulated anion channels, and P2X7
hypothesis that the enteric glia is affected by ZIKV infection in vivo, receptors (Duan et al., 2003; Ficker et al., 2014; Yang et al., 2019; Ye
as well as the possible consequences of this infection, have yet to be et al., 2003). In astrocytes, P2X7 plays a crucial role in neuroinflamma-
demonstrated. Of note, accumulating evidence has revealed that GI tion and is possibly involved in the pathophysiology of ZIKV infection
disorders are possible pathological initiators of PD many years before (Alves et al., 2020). Furthermore, astroglial permeabilization via hemi-
it progresses to the CNS, with an important role for enteric glial cells channel formation and consequent glutamate release can be stimu-
(Yang et al., 2022). lated by the pro-inflammatory cytokines, TNF-α and IL-1β (Orellana
et al., 2011), an event that may be relevant during ZIKV infection and
underlying brain damage. In line with this, the brains of convalescent
4 | ZIKV INFECTION AND POTENTIAL mice (1-year post infection) present cellular infiltration, surrounded by
N E U R O L O G I CA L C O M P L I C A T I O N S R E L A T E D activated astrocytes and microglia (Ireland et al., 2020), and the result-
TO THE A DU LT AND A G I NG B R AIN ing neuroinflammation might increase the risk of neurodegenerative
disorders. In fact, adult mice that were neonatally infected by ZIKV
Pathological processes in adult and aging brains are commonly associ- showed increased susceptibility to chemically induced seizures, neuro-
ated with the triad neuroinflammation, oxidative stress, and glutamate degeneration, and brain calcifications, which may be related to early
excitotoxicity, which are intimately related to functions of glial cells TNF-α-mediated neuroinflammation (Nem de Oliveira Souza
10981136, 2023, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/glia.24353 by UNSA - Univ Nacional de Salta, Wiley Online Library on [06/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1798 QUINCOZES-SANTOS ET AL.

et al., 2018). The role of neuroinflammation in neurocognitive impair- been observed. The phosphorylation of cAMP responsive element
ment has also been described in adult rhesus macaques infected with binding protein (CREB), an important transcription factor that medi-
ZIKV (Hsu et al., 2022). ates learning and memory processes, decreased (Figueiredo
With regard to its molecular mechanisms in the mature brain, et al., 2019). Our group recently demonstrated, in hippocampal slices,
ZIKV has been shown to affect pathways involved in synaptic plastic- which exhibit a partially preserved neuronal circuitry and active net of
ity and inflammatory response. A reduced synaptic density in the CA3 glial cells, that acute ZIKV exposure caused a marked inflammatory
and dentate gyrus of the hippocampus, accompanied by memory response. In addition, ZIKV increased the expression of TLR2 and
impairment and compromised long-term potentiation (LTP), has also adenosine receptor A2a, both of which can modulate

F I G U R E 3 Gene interaction network and functional category of top-ranked selected diseases associated with Neurological Disorders
(MalaCards DB) in the Zika infection of glial cells. (a) Astrocytes; (b) Microglia; (c) Oligodendrocytes. The networks were built using
Cytoscape v.3.8.0.
10981136, 2023, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/glia.24353 by UNSA - Univ Nacional de Salta, Wiley Online Library on [06/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
QUINCOZES-SANTOS ET AL. 1799

neuroinflammation and are involved in adult hippocampal neurogen- 5 | C O N C LU D I N G R E M A R K S


esis and synaptic plasticity. Although the expression of synaptophysin,
widely used as a marker of synaptic plasticity, was not affected by Considering the important functions of glial cells in the maintenance
short-duration ZIKV exposure, extracellular levels of brain-derived of nervous system homeostasis, as well as their roles in the pathogen-
neurotrophic factor (BDNF) and S100B were decreased, suggesting esis of neurodegenerative diseases, this review addressed functional,
that ZIKV also affects trophic signaling. Moreover, ZIKV increased cellular, and molecular mechanisms of these cells associated with
p21 senescence-associated gene expression (Bobermin et al., 2020). ZIKV infection, focusing on neurological complications in the adult
Therefore, these molecular alterations may represent a possible link and aging brain. In addition, we highlight the potential association
between ZIKV, neuroinflammation, and long-term neurological dis- between ZIKV infection of glial cells and the development and/or pro-
eases and, consequently, neurodegeneration. gression of neuropsychiatric/neurodegenerative disorders, which rep-
In addition, altered expressions of proteins previously described resent potential long-term consequences of viral infection, particularly
in several brain pathologies, such as AD, ASD, ALS, and PD, among in adult and elderly populations. It is noteworthy that epidemiological
others, have been associated with ZIKV infection (Beys-da-Silva and clinical data on long-term ZIKV are still scarce and monitoring is
et al., 2019, 2020). In fact, viral infections are one of the risk factors required, as the ZIKV epidemic is relatively recent and more concen-
for AD, and other clinically relevant neurotropic viruses (arboviruses, trated in developing countries. Finally, by understanding the glial
human immunodeficiency virus, herpesviruses, and hepatitis C virus) effects of ZIKV, preventive/therapeutic strategies focusing on glial
have previously been associated with AD (Dehhaghi et al., 2018). cells may emerge to delay and/or prevent the development of ZIKV-
With specific regard to ZIKV, infection by this virus may increase the induced neurological disorders and their future consequences.
expression of β-amyloid and phosphorylated-Tau protein, accompa-
nied by endoplasmic reticulum (ER) stress and unfolded protein AUTHOR CONTRIBU TIONS
response (UPR) in brain organoids (Lee et al., 2022). Therefore, ZIKV André Quincozes-Santos conceptualized the review. All authors wrote
infection may accelerate/aggravate clinical outcomes of neurodegen- the original draft of the manuscript, revised, edited, and approved the
erative diseases and/or age-related diseases. manuscript.
To corroborate the experimental data discussed in this review, we
performed a gene interaction network of neurological disorders asso- ACKNOWLEDG MENTS
ciated with ZIKV infection and glial cells (Figure 3). This analysis The authors are supported by the Brazilian funding agencies Conselho
reveals that ZIKV alters the expressions of genes associated with gico (CNPq); Coor-
Nacional de Desenvolvimento Científico e Tecnolo
immune/inflammatory responses, mainly in astrocytes and microglia, denação de Aperfeiçoamento Pessoal de Nível Superior (CAPES); Min-
including TNF, IL1B, IL6, TNFRSF1A, IL1R1, IFNG, TLR3, RELA, PTGS2, istry of Science, Technology, Innovation and Communications
NOS2, among others. Although the expressions of these inflammatory (MCTIC), Ministry of Education (MEC), Ministry of Health
genes can elicit an antiviral response, several of these molecules rep- (MS) through the Edital MCTIC/FNDCT-CNPq/MEC-CAPES/MS-
resent senescence markers, and chronic neuroinflammation is a com- Decit/No 14/2016, project 440763/2016-9; Fundação de Amparo à
mon feature of neuropsychiatric and neurodegenerative diseases, Pesquisa do Estado do Rio Grande do Sul (FAPERGS).
such as AD, ASD, ALS, PD, epilepsy, multiple sclerosis, and schizo-
phrenia. Accordingly, in vitro mechanistic studies have identified TNF- CONFLIC T OF INTER E ST STATEMENT
α signaling via TNFR1 as a crucial regulatory mechanism controlling The authors declare no conflicts of interest.
ZIKV-induced changes in neurologic gene expression (Kung
et al., 2022). Several genes associated with cell functionality have DATA AVAILABILITY STAT EMEN T
been identified, particularly in astrocytes, including GFAP, BDNF, Data sharing is not applicable to this article as no new data were cre-
SOD2, SLC1A3, MAOA, ACHE, APOE, LDLR, which encode astrocyte ated or analyzed in this study.
markers and proteins associated with trophic signaling, antioxidant
defenses, neurotransmitter homeostasis and cholesterol metabolism. OR CID
In oligodendrocytes, a lower number of genes were found, and these André Quincozes-Santos https://orcid.org/0000-0001-8611-4890
are associated with inflammatory processes, trophic signaling, and Larissa Daniele Bobermin https://orcid.org/0000-0001-8318-4818
metabolic processes (TNFRSF1A, NTF3, MAPK8, STAT1, FABP3). Juliana M. Coelho-Aguiar https://orcid.org/0000-0001-7451-7289
As such, the footprints left by ZIKV in glial cells may help to molec-
ularly explain the link between ZIKV and neuropsychiatric/ RE FE RE NCE S
neurodegenerative diseases. As astrocytes presented the highest num- Allaman, I., Bélanger, M., & Magistretti, P. J. (2011). Astrocyte–neuron
ber of genes associated with neurological disorders altered by ZIKV, metabolic relationships: For better and for worse. Trends in Neurosci-
ences, 34, 76–87.
these cells may represent potential key targets and cellular mediators of
Almeida, L. T., Ferraz, A. C., da Silva Caetano, C. C., da Silva
the effects of ZIKV in the CNS. These molecular analyses may also pro- Menegatto, M. B., dos Santos Pereira Andrade, A. C., Lima, R. L. S.,
vide new insights for protective strategies focused on glial cells (gliopro- Camini, F. C., Pereira, S. H., da Silva Pereira, K. Y., de Mello Silva, B.,
tective strategies) for preventing ZIKV-associated brain disorders. Perucci, L. O., Talvani, A., de Magalhães, J. C., & de Brito
10981136, 2023, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/glia.24353 by UNSA - Univ Nacional de Salta, Wiley Online Library on [06/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1800 QUINCOZES-SANTOS ET AL.

Magalhães, C. L. (2020). Zika virus induces oxidative stress and receptor blockade prevents neuronal death induced by Zika virus.
decreases antioxidant enzyme activities in vitro and in vivo. Virus Infection, mBio 8, e00350–e00317.
Research, 286, 198084. Coulter, D. A., & Eid, T. (2012). Astrocytic regulation of glutamate homeo-
Alves, V. S., Leite-Aguiar, R., da Silva, J. P., Coutinho-Silva, R., & stasis in epilepsy. Glia, 60, 1215–1226.
Savio, L. E. B. (2020). Purinergic signaling in infectious diseases of the Cumberworth, S. L., Barrie, J. A., Cunningham, M. E., de
central nervous system. Brain, Behavior, and Immunity, 89, 480–490. Figueiredo, D. P. G., Schultz, V., Wilder-Smith, A. J., Brennan, B.,
Alves-Leon, S. V., da Lima, M. R., PCG, N., LMC, C., Rabelo, K., Pena, L. J., de Oliveira, F., França, R., Linington, C., Barnett, S. C.,
Nogueira, R. M. R., de Bruycker-Nogueira, F., de Azeredo, E. L., Willison, H. J., Kohl, A., & Edgar, J. M. (2017). Zika virus tropism and
Bahia, P. R., Rueda Lopes, F. C., de Souza JPB, M., Fontes- interactions in myelinating neural cell cultures: CNS cells and myelin
Dantas, F. L., Paes, M. V., Lemos, E. R., & Santos, F. B. (2019). Zika are preferentially affected. Acta Neuropathologica Communications,
virus found in brain tissue of a multiple sclerosis patient undergoing an 5, 50.
acute disseminated encephalomyelitis-like episode. Multiple Sclerosis, de Barbosa, M. H. M., de Magalhães-Barbosa, M. C., Robaina, J. R., Prata-
25, 427–430. Barbosa, A., de de Lima, M. A. M. T., & da Cunha, A. J. L. A. (2019).
Bélanger, M., Allaman, I., & Magistretti, P. J. (2011). Brain energy metabo- Auditory findings associated with Zika virus infection: An integrative
lism: Focus on astrocyte-neuron metabolic cooperation. Cell Metabo- review. Brazilian Journal of Otorhinolaryngology, 85, 642–663.
lism, 14, 724–738. Nem de Oliveira Souza, I., Frost, P. S., França, J. V., Nascimento-
Beys-da-Silva, W. O., Quincozes-Santos, A., Tureta, E. F., Rosa, R. L., Viana, J. B., Neris, R. L. S., Freitas, L., Pinheiro, D. J. L. L.,
Berger, M., Bobermin, L. D., Souza, D. O., Guimarães, J. A., & Santi, L. Nogueira, C. O., Neves, G., Chimelli, L., De Felice, F. G.,
(2020). Association between Zika virus and future neurological dis- Cavalheiro, E. A., Ferreira, S. T., Assunção-Miranda, I., Figueiredo, C. P.,
eases. Journal of the Neurological Sciences, 409, 116617. Da Poian, A. T., & Clarke, J. R. (2018). Acute and chronic neurological
Beys-da-Silva, W. O., Rosa, R. L., Santi, L., Berger, M., Park, S. K., consequences of early-life Zika virus infection in mice. Science Transla-
Campos, A. R., Terraciano, P., Varela, A. P. M., Teixeira, T. F., tional Medicine, 10, eaar2749.
Roehe, P. M., Quincozes-Santos, A., Yates, J. R., Souza, D. O., Dehhaghi, M., Kazemi Shariat Panahi, H., & Guillemin, G. J. (2018). Micro-
Cirne-Lima, E. O., & Guimarães, J. A. (2019). Zika virus infection of organisms' footprint in neurodegenerative diseases. Frontiers in Cellular
human mesenchymal stem cells promotes differential expression of Neuroscience, 12, 466.
proteins linked to several neurological diseases. Molecular Neurobiol- Devhare, P., Meyer, K., Steele, R., Ray, R. B., & Ray, R. (2017). Zika virus
ogy, 56, 4708–4717. infection dysregulates human neural stem cell growth and inhibits dif-
Bhat, A. H., Dar, K. B., Anees, S., Zargar, M. A., Masood, A., Sofi, M. A., & ferentiation into neuroprogenitor cells. Cell Death & Disease, 8, e3106.
Ganie, S. A. (2015). Oxidative stress, mitochondrial dysfunction and Dhiman, G., Abraham, R., & Griffin, D. E. (2019). Human Schwann cells are
neurodegenerative diseases; a mechanistic insight. Biomedicine & Phar- susceptible to infection with Zika and yellow fever viruses, but not
macotherapy, 74, 101–110. dengue virus. Scientific Reports, 9, 9951.
Bido-Medina, R., Wirsich, J., Rodríguez, M., Oviedo, J., Miches, I., Bido, P., Duan, S., Anderson, C. M., Keung, E. C., Chen, Y., Chen, Y., &
Tusen, L., Stoeter, P., & Sadaghiani, S. (2018). Impact of Zika virus on Swanson, R. A. (2003). P2X7 receptor-mediated release of excitatory
adult human brain structure and functional organization. Annals of Clin- amino acids from astrocytes. The Journal of Neuroscience, 23, 1320–
ical Translational Neurology, 5, 752–762. 1328.
Bobermin, L. D., Quincozes-Santos, A., Santos, C. L., Varela, A. P. M., Ekberg, J. A. K., & St john, J. A. (2014). Crucial roles for olfactory ensheath-
Teixeira, T. F., Wartchow, K. M., Lissner, L. J., da Silva, A., ing cells and olfactory mucosal cells in the repair of damaged neural
Thomaz, N. K., Santi, L., Beys-da-Silva, W. O., Roehe, P. M., tracts: Neural regeneration using olfactory cells. The Anatomical
Sesterheim, P., Guimarães, J. A., Gonçalves, C.-A., & Souza, D. O. Record, 297, 121–128.
(2020). Zika virus exposure affects neuron-glia communication in the Enlow, W., Bordeleau, M., Piret, J., Ibáñez, F. G., Uyar, O., Venable, M.-C.,
hippocampal slices of adult rats. Scientific Reports, 10, 21604. Goyette, N., Carbonneau, J., Tremblay, M.-E., & Boivin, G. (2021).
Brison, E., Jacomy, H., Desforges, M., & Talbot, P. J. (2014). Novel treat- Microglia are involved in phagocytosis and extracellular digestion dur-
ment with neuroprotective and antiviral properties against a neuroin- ing Zika virus encephalitis in young adult immunodeficient mice. Jour-
vasive human respiratory virus. Journal of Virology, 88, 1548–1563. nal of Neuroinflammation, 18, 178.
Carricondo, F., & Romero-Go  mez, B. (2019). The cochlear spiral ganglion Ferrari-Marinho, T., De Marchi, L. R., & Caboclo, L. O. (2022). Clinical neu-
neurons: The auditory portion of the VIII nerve. The Anatomical Record, rophysiology of Zika virus encephalitis. Journal of Clinical Neurophysiol-
302, 463–471. ogy, 39, 259–264.
Carteaux, G., Maquart, M., Bedet, A., Contou, D., Brugières, P., Fourati, S., Ficker, C., Rozmer, K., Kato  , E., Ando , R. D., Schumann, L., Krügel, U.,
Cleret de Langavant, L., de Broucker, T., Brun-Buisson, C., Leparc- Franke, H., Sperlágh, B., Riedel, T., & Illes, P. (2014). Astrocyte-neuron
Goffart, I., & Mekontso, D. A. (2016). Zika virus associated with menin- interaction in the substantia gelatinosa of the spinal cord dorsal horn
goencephalitis. The New England Journal of Medicine, 374, 1595–1596. via P2X7 receptor-mediated release of glutamate and reactive oxygen
CHA, B.-M., Neto, A. M., Calvet, G. A., Kara, E. O., Lacerda, M. V. G., da species. Glia, 62, 1671–1686.
Castilho, M. C., Ströher, U., Antunes de Brito, C. A., Modjarrad, K., Figueiredo, C. P., Barros-Aragão, F. G. Q., Neris, R. L. S., Frost, P. S.,
Broutet, N., Brasil, P., Bispo de Filippis, A. M., Franca, R. F. O., & ZIKA- Soares, C., Souza, I. N. O., Zeidler, J. D., Zamberlan, D. C., de
BRA Study Team. (2019). Zika virus in rectal swab samples. Emerging Sousa, V. L., Souza, A. S., Guimarães, A. L. A., Bellio, M., Marcondes de
Infectious Diseases, 25, 951–954. Souza, J., Alves-Leon, S. V., Neves, G. A., Paula-Neto, H. A.,
Chen, J., Yang, Y., Yang, Y., Zou, P., Chen, J., He, Y., Shui, S., Cui, Y., Bai, R., Castro, N. G., De Felice, F. G., Assunção-Miranda, I., … Ferreira, S. T.
Liang, Y., Hu, Y., Jiang, B., Lu, L., Zhang, X., Liu, J., & Xu, J. (2018). AXL (2019). Zika virus replicates in adult human brain tissue and impairs
promotes Zika virus infection in astrocytes by antagonizing type I synapses and memory in mice. Nature Communications, 10, 3890.
interferon signalling. Nature Microbiology, 3, 302–309. Fontaine, K. A., Sanchez, E. L., Camarda, R., & Lagunoff, M. (2015). Dengue
Costa, V. V., Del Sarto, J. L., Rocha, R. F., Silva, F. R., Doria, J. G., virus induces and requires glycolysis for optimal replication. Journal of
Olmo, I. G., Marques, R. E., Queiroz-Junior, C. M., Foureaux, G., Virology, 89, 2358–2366.
Araújo, J. M. S., Cramer, A., Real, A. L. C. V., Ribeiro, L. S., Sardi, S. I., França, G. V. A., Schuler-Faccini, L., Oliveira, W. K., Henriques, C. M. P.,
Ferreira, A. J., Machado, F. S., de Oliveira, A. C., Teixeira, A. L., Carmo, E. H., Pedi, V. D., Nunes, M. L., Castro, M. C., Serruya, S.,
Nakaya, H. I., … Teixeira, M. M. (2017). N-methyl-d-aspartate (NMDA) Silveira, M. F., Barros, F. C., & Victora, C. G. (2016). Congenital Zika
10981136, 2023, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/glia.24353 by UNSA - Univ Nacional de Salta, Wiley Online Library on [06/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
QUINCOZES-SANTOS ET AL. 1801

virus syndrome in Brazil: A case series of the first 1501 livebirths with Jhan, M.-K., Chen, C.-L., Shen, T.-J., Tseng, P.-C., Wang, Y.-T., Satria, R. D.,
complete investigation. Lancet, 388, 891–897. Yu, C.-Y., & Lin, C.-F. (2021). Polarization of type 1 macrophages is
Gaburro, J., Bhatti, A., Sundaramoorthy, V., Dearnley, M., Green, D., associated with the severity of viral encephalitis caused by Japanese
Nahavandi, S., Paradkar, P. N., & Duchemin, J.-B. (2018). Zika virus- encephalitis virus and dengue virus. Cell, 10, 3181.
induced hyper excitation precedes death of mouse primary neuron. Jiwaji, Z., & Hardingham, G. E. (2022). Good, bad, and neglectful: Astrocyte
Virology Journal, 15, 79. changes in neurodegenerative disease. Free Radical Biology and Medi-
Garber, C., Soung, A., Vollmer, L. L., Kanmogne, M., Last, A., Brown, J., & cine, 182, 93–99.
Klein, R. S. (2019). T cells promote microglia-mediated synaptic elimi- Kung, P.-L., Chou, T.-W., Lindman, M., Chang, N. P., Estevez, I.,
nation and cognitive dysfunction during recovery from neuropatho- Buckley, B. D., Atkins, C., & Daniels, B. P. (2022). Zika virus-induced
genic flaviviruses. Nature Neuroscience, 22, 1276–1288. TNF-α signaling dysregulates expression of neurologic genes associ-
García-Marín, V., García-Lo  pez, P., & Freire, M. (2007). Cajal's contribu- ated with psychiatric disorders. Journal of Neuroinflammation, 19, 100.
tions to glia research. Trends in Neurosciences, 30, 479–487. Labzin, L. I., Heneka, M. T., & Latz, E. (2018). Innate immunity and neuro-
Gasco, S., & Muñoz-Fernández, M. Á. (2020). A review on the current degeneration. Annual Review of Medicine, 69, 437–449.
knowledge on ZIKV infection and the interest of organoids and nano- Leal, M. C., Muniz, L. F., Ferreira, T. S. A., Santos, C. M., Almeida, L. C., Van
technology on development of effective therapies against Zika infec- Der Linden, V., Ramos, R. C. F., Rodrigues, L. C., & Neto, S. S. C.
tion. IJMS, 22, 35. (2016). Hearing loss in infants with microcephaly and evidence of con-
Gershon, M. D. (2011). Behind an enteric neuron there may lie a glial cell. genital Zika virus infection — Brazil, November 2015–may 2016.
The Journal of Clinical Investigation, 121, 3386–3389. MMWR. Morbidity and Mortality Weekly Report, 65, 917–919.
Ghosh, M., Yang, Y., Rothstein, J. D., & Robinson, M. B. (2011). Nuclear Ledur, P. F., Karmirian, K., da Pedrosa, C. S. G., LRQ, S., Assis-de-
factor-B contributes to neuron-dependent induction of glutamate Lemos, G., Martins, T. M., de Ferreira, J. C. C. G., de Azevedo
transporter-1 expression in astrocytes. Journal of Neuroscience, 31, Reis, G. F., Silva, E. S., Silva, D., Salerno, J. A., Ornelas, I. M., Devalle, S.,
9159–9169. da Costa Madeiro, R. F., Goto-Silva, L., Higa, L. M., Melo, A., Tanuri, A.,
Gilbert-Jaramillo, J., Garcez, P., James, W., Molnár, Z., & Clarke, K. (2019). Chimelli, L., … Rehen, S. K. (2020). Zika virus infection leads to mito-
The potential contribution of impaired brain glucose metabolism to chondrial failure, oxidative stress and DNA damage in human iPSC-
congenital Zika syndrome. Journal of Anatomy, 235, 468–480. derived astrocytes. Scientific Reports, 10, 1218.
Gim, E., Shim, D.-W., Hwang, I., Shin, O. S., & Yu, J.-W. (2019). Zika virus Lee, S.-E., Choi, H., Shin, N., Kong, D., Kim, N. G., Kim, H.-Y., Kim, M.-J.,
impairs host NLRP3-mediated inflammasome activation in an Choi, S. W., Kim, Y. B., & Kang, K.-S. (2022). Zika virus infection accel-
NS3-dependent manner. Immune Network, 19, e40. erates Alzheimer's disease phenotypes in brain organoids. Cell Death
Greenhalgh, A. D., David, S., & Bennett, F. C. (2020). Immune cell regula- Discovery, 8, 153.
tion of glia during CNS injury and disease. Nature Reviews. Neurosci- Lewerenz, J., & Maher, P. (2015). Chronic glutamate toxicity in neurode-
ence, 21, 139–152. generative diseases—What is the evidence? Frontiers in Neurosci-
Gundersen, V., Storm-Mathisen, J., & Bergersen, L. H. (2015). Neuroglial ence,9:469. https://doi.org/10.3389/fnins.2015.00469.
transmission. Physiological Reviews, 95, 695–726. Li, C., Deng, Y.-Q., Zu, S., Quanquin, N., Shang, J., Tian, M., Ji, X.,
Henriques-Souza, A., de Azevedo Marques, E. T., Antunes de Brito, C. A., Zhang, N.-N., Dong, H.-L., Xu, Y.-P., Zhao, L.-Z., Zhang, F.-C., Li, X.-F.,
Brito Ferreira, M. L., de Morais Machado, M. Í., Pena, L. J., Wu, A., Cheng, G., & Qin, C.-F. (2018). Zika virus shedding in the stool
Cordeiro, M. T., & Moreira, Á. J. P. (2017). Guillain–Barré syndrome, and infection through the anorectal mucosa in mice. Emerging
acute disseminated encephalomyelitis and encephalitis associated with Microbes & Infections, 7, 1–10.
Zika virus infection in Brazil: Detection of viral RNA and isolation of Li, C., Wang, Q., Jiang, Y., Ye, Q., Xu, D., Gao, F., Xu, J. W., Wang, R.,
virus during late infection. The American Journal of Tropical Medicine Zhu, X., Shi, L., Yu, L., Zhang, F., Guo, W., Zhang, L., Qin, C.-F., & Xu, Z.
and Hygiene, 97, 1405–1409. (2018). Disruption of glial cell development by Zika virus contributes
Hsu, D. C., Chumpolkulwong, K., Corley, M. J., Hunsawong, T., to severe microcephalic newborn mice. Cell Discovery, 4, 43.
Inthawong, D., Schuetz, A., Imerbsin, R., Silsorn, D., Nadee, P., Li, L., Acioglu, C., Heary, R. F., & Elkabes, S. (2021). Role of astroglial toll-
Sopanaporn, J., Phuang-Ngern, Y., Klungthong, C., Reed, M., like receptors (TLRs) in central nervous system infections, injury and
Fernandez, S., Ndhlovu, L. C., Paul, R., Lugo-Roman, L., Michael, N. L., neurodegenerative diseases. Brain, Behavior, and Immunity, 91,
Modjarrad, K., & Vasan, S. (2022). Neurocognitive impact of Zika virus 740–755.
infection in adult rhesus macaques. Journal of Neuroinflammation, Liang, Q., Luo, Z., Zeng, J., Chen, W., Foo, S.-S., Lee, S.-A., Ge, J., Wang, S.,
19, 40. Goldman, S. A., Zlokovic, B. V., Zhao, Z., & Jung, J. U. (2016). Zika virus
Huang, Y., Li, Y., Zhang, H., Zhao, R., Jing, R., Xu, Y., He, M., Peer, J., NS4A and NS4B proteins deregulate Akt-mTOR signaling in human
Kim, Y. C., Luo, J., Tong, Z., & Zheng, J. (2018). Zika virus propagation fetal neural stem cells to inhibit neurogenesis and induce autophagy.
and release in human fetal astrocytes can be suppressed by neutral Cell Stem Cell, 19, 663–671.
sphingomyelinase-2 inhibitor GW4869. Cell Discovery, 4, 19. Lima, M. C., de Mendonça, L. R., Rezende, A. M., Carrera, R. M., Aníbal-
Hughes, M. M., & O'Neill, L. A. J. (2018). Metabolic regulation of NLRP3. Silva, C. E., Demers, M., D'Aiuto, L., Wood, J., Chowdari, K. V.,
Immunological Reviews, 281, 88–98. Griffiths, M., Lucena-Araujo, A. R., Barral-Netto, M., Azevedo, E. A. N.,
Inglis, F. M., Lee, K. M., Chiu, K. B., Purcell, O. M., Didier, P. J., Russell- Alves, R. W., Farias, P. C. S., Marques, E. T. A., Castanha, P. M. S.,
Lodrigue, K., Weaver, S. C., Roy, C. J., & MacLean, A. G. (2016). Neuro- Donald, C. L., Kohl, A., … Franca, R. F. O. (2019). The transcriptional
pathogenesis of chikungunya infection: Astrogliosis and innate and protein profile from human infected neuroprogenitor cells is
immune activation. Journal of Neurovirology, 22, 140–148. strongly correlated to Zika virus microcephaly cytokines phenotype
Ireland, D. D. C., Manangeeswaran, M., Lewkowicz, A. P., Engel, K., evidencing a persistent inflammation in the CNS. Frontiers in Immunol-
Clark, S. M., Laniyan, A., Sykes, J., Lee, H.-N., McWilliams, I. L., Kelley- ogy, 10, 1928.
Baker, L., Tonelli, L. H., & Verthelyi, D. (2020). Long-term persistence Lindqvist, R., Mundt, F., Gilthorpe, J. D., Wölfel, S., Gekara, N. O.,
of infectious Zika virus: Inflammation and behavioral sequela in mice. Kröger, A., & Överby, A. K. (2016). Fast type I interferon response pro-
PLoS Pathogens, 16, e1008689. tects astrocytes from flavivirus infection and virus-induced cytopathic
Jäkel, S., & Dimou, L. (2017). Glial cells and their function in the adult effects. Journal of Neuroinflammation, 13, 277.
brain: A journey through the history of their ablation. Frontiers in Cell Lum, F.-M., Low, D. K. S., Fan, Y., Tan, J. J. L., Lee, B., Chan, J. K. Y.,
Neuroscience, 11. https://doi.org/10.3389/fncel.2017.00024 Rénia, L., Ginhoux, F., & Ng, L. F. P. (2017). Zika virus infects human
10981136, 2023, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/glia.24353 by UNSA - Univ Nacional de Salta, Wiley Online Library on [06/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1802 QUINCOZES-SANTOS ET AL.

fetal brain microglia and induces inflammation. Clinical Infectious Dis- Palsson-McDermott, E. M., Curtis, A. M., Goel, G., Lauterbach, M. A. R.,
eases, 64, 914–920. Sheedy, F. J., Gleeson, L. E., van den Bosch, M. W. M., Quinn, S. R.,
Lynch, M. A. (2020). Can the emerging field of immunometabolism provide Domingo-Fernandez, R., Johnston, D. G. W., Jiang, J., Israelsen, W. J.,
insights into neuroinflammation? Progress in Neurobiology, 184, Keane, J., Thomas, C., Clish, C., Vander Heiden, M., Xavier, R. J., &
101719. O'Neill, L. A. J. (2015). Pyruvate kinase M2 regulates Hif-1α activity
Mai, J. K., & Paxinos, G. (Eds.). (2012). The human nervous system (3rd ed.). and IL-1β induction and is a critical determinant of the Warburg effect
Elsevier Academic Press. in LPS-activated macrophages. Cell Metabolism, 21, 65–80.
Martínez, L. E., Garcia, G., Contreras, D., Gong, D., Sun, R., & Petersen, L. R., Jamieson, D. J., Powers, A. M., & Honein, M. A. (2016). Zika
Arumugaswami, V. (2020). Zika virus mucosal infection provides pro- virus. The New England Journal of Medicine, 374, 1552–1563.
tective immunity. Journal of Virology, 94, e00067–e00020. Quincozes-Santos, A., Santos, C. L., de Souza Almeida, R. R., da Silva, A.,
Mattson, M. P., & Arumugam, T. V. (2018). Hallmarks of brain aging: Adap- Thomaz, N. K., Costa, N. L. F., Weber, F. B., Schmitz, I., Medeiros, L. S.,
tive and pathological modification by metabolic states. Cell Metabolism, Medeiros, L., Dotto, B. S., Dias, F. R. P., Sovrani, V., & Bobermin, L. D.
27, 1176–1199. (2021). Gliotoxicity and glioprotection: The dual role of glial cells.
Maucourant, C., Queiroz, G. A. N., Samri, A., Grassi, M. F. R., Yssel, H., & Molecular Neurobiology, 58, 6577–6592.
Vieillard, V. (2019). Zika virus in the eye of the cytokine storm. Rawal, G., Yadav, S., & Kumar, R. (2016). Zika virus: An overview. Journal
European Cytokine Network, 30, 74–81. of Family Medicine and Primary Care, 5, 523–527.
Mauro, C., Leow, S. C., Anso, E., Rocha, S., Thotakura, A. K., Tornatore, L., Retallack, H., Di Lullo, E., Arias, C., Knopp, K. A., Laurie, M. T., Sandoval-
Moretti, M., De Smaele, E., Beg, A. A., Tergaonkar, V., Chandel, N. S., & Espinosa, C., Mancia Leon, W. R., Krencik, R., Ullian, E. M.,
Franzoso, G. (2011). NF-κB controls energy homeostasis and meta- Spatazza, J., Pollen, A. A., Mandel-Brehm, C., Nowakowski, T. J.,
bolic adaptation by upregulating mitochondrial respiration. Nature Cell Kriegstein, A. R., & DeRisi, J. L. (2016). Zika virus cell tropism in the
Biology, 13, 1272–1279. developing human brain and inhibition by Azithromycin. Proceedings
Meertens, L., Labeau, A., Dejarnac, O., Cipriani, S., Sinigaglia, L., Bonnet- of the National Academy of Sciences of the United States of America,
Madin, L., Le Charpentier, T., Hafirassou, M. L., Zamborlini, A., Cao- 113, 14408–14413.
Lormeau, V.-M., Coulpier, M., Missé, D., Jouvenet, N., Tabibiazar, R., Rombi, F., Bayliss, R., Tuplin, A., & Yeoh, S. (2020). The journey of Zika to
Gressens, P., Schwartz, O., & Amara, A. (2017). Axl mediates ZIKA the developing brain. Molecular Biology Reports, 47, 3097–3115.
virus entry in human glial cells and modulates innate immune Rossi, F., Josey, B., Sayitoglu, E. C., Potens, R., Sultu, T., Duru, A. D., &
responses. Cell Reports, 18, 324–333. Beljanski, V. (2020). Characterization of Zika virus infection of human
Mehta, A., Prabhakar, M., Kumar, P., Deshmukh, R., & Sharma, P. L. (2013). fetal cardiac mesenchymal stromal cells. PLoS One, 15, e0239238.
Excitotoxicity: Bridge to various triggers in neurodegenerative disor- Rudy, C. C., Hunsberger, H. C., Weitzner, D. S., & Reed, M. N. (2015). The
ders. European Journal of Pharmacology, 698, 6–18. role of the tripartite glutamatergic synapse in the pathophysiology of
Mlakar, J., Korva, M., Tul, N., Popovic, M., Poljšak-Prijatelj, M., Mraz, J., Alzheimer's disease. Aging and Disease, 6, 131–148.
Kolenc, M., Resman Rus, K., Vesnaver Vipotnik, T., Fabjan Vodušek, V., Ryan, F. J., Carr, J. M., Furtado, J. M., Ma, Y., Ashander, L. M., Simões, M.,
Vizjak, A., Pižem, J., Petrovec, M., & Avšič, Ž. T. (2016). Zika virus asso- Oliver, G. F., Granado, G. B., Dawson, A. C., Michael, M. Z.,
ciated with microcephaly. The New England Journal of Medicine, 374, Appukuttan, B., Lynn, D. J., & Smith, J. R. (2021). Zika virus infection
951–958. of human iris pigment epithelial cells. Frontiers in Immunology, 12,
Molina-Gonzalez, I., Miron, V. E., & Antel, J. P. (2022). Chronic oligoden- 644153.
drocyte injury in central nervous system pathologies. Communications Santi, L., Riesgo, R. S., Quincozes-Santos, A., Schuler-Faccini, L.,
Biology, 5, 1274. Tureta, E. F., Rosa, R. L., Berger, M., Oliveira, A. C. C., Beltrão-
Musso, D., Ko, A. I., & Baud, D. (2019). Zika virus infection — After the Braga, P. C. B., Souza, D. O., Guimarães, J. A., & Beys-da-Silva, W. O.
pandemic. The New England Journal of Medicine, 381, 1444–1457. (2021). Zika virus infection associated with autism spectrum disorder:
Mutso, M., St John, J. A., Ling, Z. L., Burt, F. J., Poo, Y. S., Liu, X., A case report. Neuroimmunomodulation, 28, 229–232.
Žusinaite, E., Grau, G. E., Hueston, L., Merits, A., King, N. J. C., Schouest, B., Peterson, T. A., Szeltner, D. M., Scheef, E. A., Baddoo, M.,
Ekberg, J. A. K., & Mahalingam, S. (2020). Basic insights into Zika virus Ungerleider, N., Flemington, E. K., MacLean, A. G., & Maness, N. J.
infection of neuroglial and brain endothelial cells. Journal of General (2021). Transcriptional signatures of Zika virus infection in astrocytes.
Virology, 101, 622–634. Journal of Neurovirology, 27, 116–125.
Oh, Y., Zhang, F., Wang, Y., Lee, E. M., Choi, I. Y., Lim, H., Mirakhori, F., Schreiner, B., Romanelli, E., Liberski, P., Ingold-Heppner, B., Sobottka-
Li, R., Huang, L., Xu, T., Wu, H., Li, C., Qin, C.-F., Wen, Z., Wu, Q.-F., Brillout, B., Hartwig, T., Chandrasekar, V., Johannssen, H.,
Tang, H., Xu, Z., Jin, P., Song, H., … Lee, G. (2017). Zika virus directly Zeilhofer, H. U., Aguzzi, A., Heppner, F., Kerschensteiner, M., &
infects peripheral neurons and induces cell death. Nature Neuroscience, Becher, B. (2015). Astrocyte depletion impairs redox homeostasis and
20, 1209–1212. triggers neuronal loss in the adult CNS. Cell Reports, 12, 1377–1384.
Ojha, C. R., Rodriguez, M., Karuppan, M. K. M., Lapierre, J., Schuler-Faccini, L., del Campo, M., García-Alix, A., Ventura, L. O.,
Kashanchi, F., & El-Hage, N. (2019). Toll-like receptor 3 regulates Zika Boquett, J. A., van der Linden, V., Pessoa, A., van der Linden, J. H.,
virus infection and associated host inflammatory response in primary Ventura, C. V., Leal, M. C., Kowalski, T. W., Rodrigues Gerzson, L.,
human astrocytes. PLoS One, 14, e0208543. Skilhan de Almeida, C., Santi, L., Beys-da-Silva, W. O., Quincozes-
Olmo, I. G., Carvalho, T. G., Costa, V. V., Alves-Silva, J., Ferrari, C. Z., Santos, A., Guimarães, J. A., Garcez, P. P., do Gomes, J. A., …
Izidoro-Toledo, T. C., da Silva, J. F., Teixeira, A. L., Souza, D. G., Souza, D. O. (2022). Neurodevelopment in children exposed to Zika in
Marques, J. T., Teixeira, M. M., Vieira, L. B., & Ribeiro, F. M. (2017). utero: Clinical and molecular aspects. Frontiers in Genetics, 13, 758715.
Zika virus promotes neuronal cell death in a non-cell autonomous Schultz, V., Barrie, J. A., Donald, C. L., Crawford, C. L., Mullin, M.,
manner by triggering the release of neurotoxic factors. Frontiers in Anderson, T. J., Solomon, T., Barnett, S. C., Linington, C., Kohl, A.,
Immunology, 8, 1016. Willison, H. J., & Edgar, J. M. (2021). Oligodendrocytes are susceptible
Orellana, J. A., Froger, N., Ezan, P., Jiang, J. X., Bennett, M. V. L., to Zika virus infection in a mouse model of perinatal exposure: Implica-
Naus, C. C., Giaume, C., & Sáez, J. C. (2011). ATP and glutamate tions for CNS complications. Glia, 69, 2023–2036.
released via astroglial connexin 43 hemichannels mediate neuronal Schultz, V., Cumberworth, S. L., Gu, Q., Johnson, N., Donald, C. L.,
death through activation of pannexin 1 hemichannels. Journal of Neu- McCanney, G. A., Barrie, J. A., Da Silva, F. A., Linington, C.,
rochemistry, 118, 826–840. Willison, H. J., Edgar, J. M., Barnett, S. C., & Kohl, A. (2021). Zika virus
10981136, 2023, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/glia.24353 by UNSA - Univ Nacional de Salta, Wiley Online Library on [06/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
QUINCOZES-SANTOS ET AL. 1803

infection leads to demyelination and axonal injury in mature CNS cul- Vianna, P., do Gomes, J. A., Boquett, J. A., Fraga, L. R., Schuch, J. B.,
tures. Viruses, 13, 91. FSL, V., & Schuler-Faccini, L. (2018). Zika virus as a possible risk factor
Shad, K. F., Soubra, W., & Cordato, D. J. (2022). The auditory afferent for autism spectrum disorder: Neuroimmunological aspects. Neuroim-
pathway as a clinical marker of Alzheimer's disease. JAD, 85, 47–53. munomodulation, 25, 320–327.
Shang, J., Li, C., Jin, Z., Zu, S., Chen, S., Chen, J., Chen, Z., Tang, H., Vizuete, A. F. K., Fro  es, F., Seady, M., Zanotto, C., Bobermin, L. D.,
Qin, C.-F., Ye, Q., & Wu, A. (2022). Immune profiles in mouse brain Roginski, A. C., Wajner, M., Quincozes-Santos, A., & Gonçalves, C. A.
and testes infected by Zika virus with variable pathogenicity. Frontiers (2022). Early effects of LPS-induced neuroinflammation on the rat hip-
in Cellular and Infection Microbiology, 12, 948980. pocampal glycolytic pathway. Journal of Neuroinflammation, 19, 255.
Shereen, M. A., Bashir, N., Su, R., Liu, F., Wu, K., Luo, Z., & Wu, J. (2021). Volpi, V. G., Pagani, I., Ghezzi, S., Iannacone, M., D'Antonio, M., &
Zika virus dysregulates the expression of astrocytic genes involved Vicenzi, E. (2018). Zika virus replication in dorsal root ganglia explants
in neurodevelopment. PLoS Neglected Tropical Diseases, 15, from interferon Receptor1 knockout mice causes myelin degeneration.
e0009362. Scientific Reports, 8, 10166.
Sirohi, D., & Kuhn, R. J. (2017). Can an FDA-approved Alzheimer's drug be Wang, J., Liu, J., Zhou, R., Ding, X., Zhang, Q., Zhang, C., & Li, L. (2018).
repurposed for alleviating neuronal symptoms of Zika virus? MBio, 8, Zika virus infected primary microglia impairs NPCs proliferation and
1–6. differentiation. Biochemical and Biophysical Research Communications,
St. John, J. A., Walkden, H., Nazareth, L., Beagley, K. W., Ulett, G. C., 497, 619–625.
Batzloff, M. R., Beacham, I. R., & Ekberg, J. A. K. (2016). Burkholderia Wu, S., Zeng, Y., Lerner, A., Gao, B., & Law, M. (2018). Nervous system
pseudomallei rapidly infects the brain stem and spinal cord via the tri- injury and neuroimaging of Zika virus infection. Frontiers in Neurology,
geminal nerve after intranasal inoculation. Infection and Immunity, 84, 9, 227.
2681–2688. Yang, H., Li, S., & Le, W. (2022). Intestinal permeability, dysbiosis, inflam-
Stefanik, M., Formanova, P., Bily, T., Vancova, M., Eyer, L., Palus, M., mation and enteric glia cells: The intestinal etiology of Parkinson's dis-
Salat, J., Braconi, C. T., de Zanotto, P. M. A., Gould, E. A., & Ruzek, D. ease. Aging and Disease, 13, 1381–1390.
(2018). Characterisation of Zika virus infection in primary human Yang, J., Vitery, M. D. C., Chen, J., Osei-Owusu, J., Chu, J., & Qiu, Z.
astrocytes. BMC Neuroscience, 19, 5. (2019). Glutamate-releasing SWELL1 channel in astrocytes modulates
Tricarico, P. M., Caracciolo, I., Crovella, S., & D'Agaro, P. (2017). Zika synaptic transmission and promotes brain damage in stroke. Neuron,
virus induces inflammasome activation in the glial cell line U87-MG. 102, 813–827.e6.
Biochemical and Biophysical Research Communications, 492, Ye, Z.-C., Wyeth, M. S., Baltan-Tekkok, S., & Ransom, B. R. (2003). Func-
597–602. tional hemichannels in astrocytes: A novel mechanism of glutamate
Ubeda-Bañon, I., Saiz-Sanchez, D., Flores-Cuadrado, A., Rioja-Corroto, E., release. The Journal of Neuroscience, 23, 3588–3596.
Gonzalez-Rodriguez, M., Villar-Conde, S., Astillero-Lopez, V., Cabello- Zafar, M. I., Yu, J., & Li, H. (2021). Implications of RNA viruses in the male
de la Rosa, J. P., Gallardo-Alcañiz, M. J., Vaamonde-Gamo, J., Relea- reproductive tract: An outlook on SARS-CoV-2. Frontiers in Microbiol-
Calatayud, F., Gonzalez-Lopez, L., Mohedano-Moriano, A., ogy, 12, 783963.
Rabano, A., & Martinez-Marcos, A. (2020). The human olfactory sys- Zhang, F., Wang, H.-J., Wang, Q., Liu, Z.-Y., Yuan, L., Huang, X.-Y., Li, G.,
tem in two proteinopathies: Alzheimer's and Parkinson's diseases. Ye, Q., Yang, H., Shi, L., Deng, Y.-Q., Qin, C.-F., & Xu, Z. (2017). Ameri-
Translational Neurodegeneration, 9, 22. can strain of Zika virus causes more severe microcephaly than an old
Vainchtein, I. D., & Molofsky, A. V. (2020). Astrocytes and microglia: In Asian strain in neonatal mice. eBioMedicine, 25, 95–105.
sickness and in health. Trends in Neurosciences, 43, 144–154. Zorec, R., Parpura, V., & Verkhratsky, A. (2018). Astroglial vesicular net-
Vardjan, N., Parpura, V., Verkhratsky, A., & Zorec, R. (2019). Gliocrine sys- work: Evolutionary trends, physiology and pathophysiology. Acta Phy-
tem: Astroglia as secretory cells of the CNS. Advances in Experimental siologica, 222(2). https://doi.org/10.1111/apha.12915.
Medicine and Biology, 1175, 93–115.
Vardjan, N., & Zorec, R. (2015). Excitable astrocytes: Ca2+ and cAMP-
regulated exocytosis. Neurochemical Research, 40, 2414–2424.
Veríssimo, C. P., da Carvalho, J. S., da Silva, F. J. M., Campanati, L., Moura- How to cite this article: Quincozes-Santos, A., Bobermin,
Neto, V., & de Coelho-Aguiar, J. M. (2019). Laminin and environmental
L. D., Costa, N. L. F., Thomaz, N. K., Almeida, R. R. d. S.,
cues act in the inhibition of the neuronal differentiation of enteric glia
in vitro. Frontiers in Neuroscience, 13, 914. Beys-da-Silva, W. O., Santi, L., Rosa, R. L., Capra, D.,
Verkhratsky, A., Ho, M. S., Zorec, R., & Parpura, V. (2019). The concept of Coelho-Aguiar, J. M., DosSantos, M. F., Heringer, M.,
neuroglia. In A. Verkhratsky, M. S. Ho, R. Zorec, & V. Parpura (Eds.), Cirne-Lima, E. O., Guimarães, J. A., Schuler-Faccini, L.,
Neuroglia in neurodegenerative diseases. Advances in experimental medi-
Gonçalves, C.-A., Moura-Neto, V., & Souza, D. O. (2023). The
cine and biology (Vol. 1175, pp. 1–13). Springer Singapore.
Verma, M., Lizama, B. N., & Chu, C. T. (2022). Excitotoxicity, calcium and role of glial cells in Zika virus-induced neurodegeneration. Glia,
mitochondria: A triad in synaptic neurodegeneration. Translational 71(8), 1791–1803. https://doi.org/10.1002/glia.24353
Neurodegeneration, 11, 3.

Você também pode gostar